WO2016179252A1 - Sprinkle formulations of acamprosate - Google Patents

Sprinkle formulations of acamprosate Download PDF

Info

Publication number
WO2016179252A1
WO2016179252A1 PCT/US2016/030725 US2016030725W WO2016179252A1 WO 2016179252 A1 WO2016179252 A1 WO 2016179252A1 US 2016030725 W US2016030725 W US 2016030725W WO 2016179252 A1 WO2016179252 A1 WO 2016179252A1
Authority
WO
WIPO (PCT)
Prior art keywords
pharmaceutical formulation
sprinkles
acamprosate
disease
acamprosate calcium
Prior art date
Application number
PCT/US2016/030725
Other languages
English (en)
French (fr)
Inventor
Steven L. JOHNS
Kenneth G. PAYIE
Badrinath R. DONIPARTHI
Shivaraj B. MUNIANJANAPPA
Original Assignee
Confluence Pharmaceuticals, Llc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Confluence Pharmaceuticals, Llc filed Critical Confluence Pharmaceuticals, Llc
Priority to US15/569,937 priority Critical patent/US10709668B2/en
Priority to SG11201708393UA priority patent/SG11201708393UA/en
Priority to KR1020177033137A priority patent/KR20170142180A/ko
Priority to JP2017558372A priority patent/JP7026347B2/ja
Priority to EP16789996.2A priority patent/EP3291800A4/en
Priority to AU2016258624A priority patent/AU2016258624B2/en
Priority to IL255343A priority patent/IL255343B1/en
Publication of WO2016179252A1 publication Critical patent/WO2016179252A1/en
Priority to ZA2017/08037A priority patent/ZA201708037B/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • A61K9/0056Mouth soluble or dispersible forms; Suckable, eatable, chewable coherent forms; Forms rapidly disintegrating in the mouth; Lozenges; Lollipops; Bite capsules; Baked products; Baits or other oral forms for animals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5073Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals having two or more different coatings optionally including drug-containing subcoatings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0087Galenical forms not covered by A61K9/02 - A61K9/7023
    • A61K9/009Sachets, pouches characterised by the material or function of the envelope
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1629Organic macromolecular compounds
    • A61K9/1652Polysaccharides, e.g. alginate, cellulose derivatives; Cyclodextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4808Preparations in capsules, e.g. of gelatin, of chocolate characterised by the form of the capsule or the structure of the filling; Capsules containing small tablets; Capsules with outer layer for immediate drug release
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5005Wall or coating material
    • A61K9/5021Organic macromolecular compounds
    • A61K9/5026Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5005Wall or coating material
    • A61K9/5021Organic macromolecular compounds
    • A61K9/5036Polysaccharides, e.g. gums, alginate; Cyclodextrin
    • A61K9/5042Cellulose; Cellulose derivatives, e.g. phthalate or acetate succinate esters of hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5005Wall or coating material
    • A61K9/5021Organic macromolecular compounds
    • A61K9/5036Polysaccharides, e.g. gums, alginate; Cyclodextrin
    • A61K9/5042Cellulose; Cellulose derivatives, e.g. phthalate or acetate succinate esters of hydroxypropyl methylcellulose
    • A61K9/5047Cellulose ethers containing no ester groups, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4866Organic macromolecular compounds

Definitions

  • the present invention relates to sprinkle formulations of acamprosate pellets and their use as a medicament.
  • NMDA N-methyl-D-aspartate
  • acamprosate may interact with excitatory glutamatergic neurotransmission in general, and as an antagonist of the metabotropic glulamate receptor subtype 5 (mGluR5) in particular (De Witte et al., CNS Dntgs2005, 19(6), 517-37).
  • mGluR5 metabotropic glulamate receptor subtype 5
  • the glutamatergic mechanism of action of acamprosate may explain the effects of acamprosate on alcohol dependence, and suggests other therapeutic activities, such as in neuroprotection.
  • acamprosate as a medicament.
  • US patent nos. 6,391,922 and 6,689,816 disclose a novel treatment for neuropsychiatric disorders, including anxiety disorders, mood disorders, psychotic disorders, somatoform disorders, and neuropsychiatric symptoms resulting from movement disorders.
  • the treatment utilizes any agent that simultaneously act as NMDA-type glutamate receptor antagonists and GABA-A receptor agonists.
  • these two activities are characteristic of a single agent, for example, acamprosate (calcium N- acetylhomotaurinate).
  • acamprosate calcium N- acetylhomotaurinate
  • separate agents having these activities can be combined as a compound or mixture and thereby administered together.
  • the invention also provides for a third agent that acts as a non-competitive NMDA-receptor blocking agent or ion channel blocker that augments the effect of the primary treatment.
  • a particularly preferred ion channel blocking agent is magnesium.
  • US patent no. 7,745,493 discloses a novel treatment for movement disorders, including tardive dyskinesia, tic disorders, Tourette's syndrome, and blepharospasm, and other focal dystonias.
  • the treatment utilizes agents that simultaneously act as NMDA-type glutamate receptor antagonists and GABA-A receptor agonists. Preferably, these two activities are characteristic of a single agent, for example acamprosate. Alternatively, separate agents having these activities can be combined and administered together.
  • the invention also provides a third agent that acts as a non-competitive NMDA-receptor blocking agent or ion channel blocker that augments the effect of the primary treatment.
  • a particularly preferred ion channel blocking agent is magnesium. Alternatively, magnesium can be administered alone for prevention and treatment of movement disorders.
  • US patent no. 8,865,769 discloses combinations and methods for the treatment of neurological disorders related to glutamate excitotoxicity and Amyloid ⁇ toxicity. More specifically, the invention relates to novel combinatorial therapies of Multiple Sclerosis, Alzheimer's disease, Alzheimer's disease related disorder, Amyotrophic Lateral Sclerosis, Parkinson's disease, Huntington's disease, neuropathic pain, alcoholic neuropathy, alcoholism or alcohol withdrawal, or spinal cord injury, based on Baclofen and Acamprosate combination.
  • WO 2010093859 Al relates to subjects who were diagnosed with either comorbid or idiopathic autism and fragile x syndrome and treated with acamprosate. Patients generally showed marked improvements in primary outcomes as assessed using, for example, standard clinic measures for functionality including the Clinical Global Impressions Improvement (CGI-I) and the Clinical Global Impressions Severity (CGI-S) scales.
  • CGI-I Clinical Global Impressions Improvement
  • CGI-S Clinical Global Impressions Severity
  • US patent no. 6,426,087 discloses an orally administrable galenic form allowing improved absorption by the transmembrane or paracellular route in the gastrointestinal tract of active ingredients which are hydrophilic or ionizable in physio! .
  • a controlled- release tablet contained ( 1) cores contg.
  • US patent no. 6,512,009 discloses a pharmaceutical composition for the treatment of alcohol and drug dependence, comprising a therapeutically effective amount of a combination of: (i) an opioid antagonist; and (ii) a NMDA receptor complex modulator.
  • a pharmaceutical kit is also provided, comprising these two substances.
  • the opioid antagonist can, for example, be naltrexone and the NMDA receptor complex modulator can be a spermidine site modulator such as acamprosate.
  • US patent no. 7,994,218 discloses pantoic acid ester neopentyl sulfonyl ester prodrugs of acamprosate, pharmaceutical compositions comprising such prodrugs, and methods of using such prodrugs and compositions thereof for treating diseases.
  • acamprosate prodrugs exhibiting enhanced oral bioavailability and methods of using acamprosate prodrugs to treat neurodegenerative disorders, psychotic disorders, mood disorders, anxiety disorders, somatoform disorders, movement disorders, substance abuse disorders, binge eating disorder, cortical spreading depression related disorders, tinnitus, sleeping disorders, multiple sclerosis, and pain are disclosed.
  • US patent no. 8,268,352 discloses a novel modified release dosage form comprising of a high solubility active ingredient, which utilizes dual retard technique to effectively reduce the quantity of release controlling agents.
  • the dosage form can optionally comprise another active ingredient as an immediate release form or modified release form.
  • the invention also relates to a process for preparing the said formulation.
  • US patent no. 9,000,046 discloses gastric retentive dosage forms for sustained release of acamprosate which may allow once- or twice-daily dosing for both acute and long-term treatment of a disorder including alcohol dependence, tinnitus, sleep apnea, Parkinson's disease, levodopa-induced dyskinesias in Parkinson's disease, Alzheimer's disease, Huntington's disease, Amyotrophic lateral sclerosis, Cortical spreading depression, migraine, schizophrenia, anxiety, tardive dyskinesia, spasticity, multiple sclerosis, various types pain, or binge eating. Methods of treatment using the dosage forms and methods of making the dosage forms are also described.
  • Acamprosate is a polar molecule that lacks the requisite physicochemical characteristics for effective passive permeability across cellular membranes.
  • the oral bioavailability of acamprosate in humans is only about 1 1 %, and poor absorption of the drug from the GI tract likely contributes to its limited lolerability.
  • One consequence is that a relatively large tablet has been required to achieve a therapeutic effect.
  • Campral® Acamprosate calcium, marketed as Campral® by Forest Pharma, was first approved by the FDA in 2004. Campral® is indicated for the maintenance of abstinence from alcohol in patients with alcohol dependence who are abstinent at treatment initiation. Campral® is supplied as an enteric-coated tablet for oral administration. Each Campral® tablet contains acamprosate calcium 333 mg, equivalent to 300 mg of acamprosate. Inactive ingredients in Campral® tablets include: crospovidone, microcrystalline cellulose, magnesium silicate, sodium starch glycolale, colloidal anhydrous silica, magnesium stearate, talc, propylene glycol and Eudragit® L 30 D or equivalent.
  • Campral® 333 mg tablets are enteric-coated, white, round, biconvex tablets, identified with "333" debossed on one side.
  • the recommended dose of Campral® is two 333 mg tablets (each dose should total 666 mg) taken three times daily. A lower dose may be effective in some patients.
  • dosing may be done without regard to meals, dosing with meals was employed during clinical trials and is suggested in those patients who regularly eat three meals daily.
  • any disruption of the coaling allows for immediate dissolution of the tablet before moving through the upper digestive tract and into the lower digestive tract for absorption.
  • Gl distress diarrhea nausea and vomiting
  • Campral® is a solid formed, 333 mg round tablet and is a 10 mm in size. Also known as AOTAL in Europe, the drug is available in 400 mg and 800 mg tablets. The size of the tablet presents a challenge for both pediatric and adult patients.
  • the FDA issued draft guidance on size, shape and other physical attributes of generic tablets and capsules which outlines the difficulties swallowing tablets and capsules for many individuals and can lead to a variety of adverse events and patient noncompliance with treatment regimens .
  • Campral® acamprosate calcium
  • FXS Fragile X Syndrome
  • CGI-I Clinical Global Impression
  • Gastrointestinal distress (nausea and vomiting) are commonly observed side effects in subjects receiving acamprosate, and adverse GI events were observed in the majority of Fragile X subjects treated in this study.
  • Figure 1 shows a dissolution profile of acamprosate calcium enteric-coated sprinkles (#25/30).
  • Figure 2 shows a dissolution profile of open petri plate study for sprinkles with 5% ethyl cellulose + 40% enteric coating.
  • Figure 3 shows a Dissolution profile of open petri plate study for sprinkles with 10% ethyl cellulose + 40% enteric coating.
  • Figure 4 shows a Dissolution profile of open petri plate study for sprinkles with 10% ethyl cellulose + 30% enteric coating.
  • Figure 5 shows a dissolution profile of Acamprosate Calcium Sprinkles (5% ethyl cellulose + 40% eudragit coating).
  • Figure 6 shows a dissolution profile of Acamprosate Calcium Sprinkles (5% ethyl cellulose + 50% eudragit coating).
  • Figure 7 shows day I PK during the first 12 hour period.
  • Figure 8 shows the day 1 PK for the 666 mg BID study.
  • Figure 9 shows PK- 1332 mg (666 mg BID) Day 1 -Campral vs. SF 15000679 vs. SF15000775 (left) compared to Campral vs. SF 14000999 (right).
  • Figure 10 shows PK- 1332 mg (666 mg BID) Day 1 vs. Day 4 ⁇ Campral vs.
  • the present invention provides an orally-administrable, pharmaceutical formulation comprising: a plurality of sprinkles, wherein:
  • the sprinkles comprise a core, a sustained release coating, and an enteric coating; and the core comprises an active ingredient and a diluent.
  • the active ingredient is a homotaurine analog selected from the group consisting of: an acetyl aminopropane sulfonate, an acetylaminopropane sulfonate salt, taltrimide, and tauromustine.
  • the active ingredient is 3 -acetamidopropane-1 -sulfonic acid or a pharmaceutically acceptable salt thereof.
  • the active ingredient is acamprosate calcium.
  • the diluent comprises Microcrystalline Cellulose (MCC) or a cellulose gel.
  • the diluent consists essentially of Microcrystalline Cellillose (kCC).
  • the diluent comprises Avicel PHI 01.
  • the diluent consists essentially of Avicel PHI 01.
  • the sustained release coating comprises a thermoplastic cellulose ether.
  • the sustained release coating comprises ethyl cellulose. In a preferred embodiment, the sustained release coating comprises Ethyl cellulose 20 standard.
  • the sustained release coating further comprises triethyl citrate.
  • the ratio of Ethyl cellulose 20 standard: triethyl citrate is about 5 to about 30 w/w%.
  • the ratio of Ethyl cellulose 20 standard: triethyl citrate is about 25 w/w%.
  • the enteric coating comprises an anionic copolymer based on meth acrylic acid and ethyl acrylate.
  • the enteric coating comprises a Eudragit.
  • the enteric coating comprises a Eudragit S.
  • the enteric coating comprises a Eudragit L.
  • the enteric coaling comprises Eudragit LI 00 55.
  • the enteric coating further comprises triethyl citrate.
  • the enteric coating further comprises talc.
  • the enteric coating further comprises triethyl citrate and talc.
  • the ratio of Eudragit LI 00 55: trielhyl citrate: talc is about 100 to about 25 to about 50 w/w%.
  • the ratio of Eudragit L I 00 55: triethyl citrate: talc is about 100 to about 25 to about 50 w/w%.
  • the plurality of pellets range in size from about 0.5 mm to about
  • the plurality of pellets range in size from about 0.6 mm to about 1.5 mm, as measured using an ASTM Mesh.
  • the active ingredient is homogeneously dispersed in the pellets.
  • the surface of the core is substantially smooth in texture.
  • the surface of the core is substantially non-porous.
  • the formulation is a sustained release formulation of acamprosate calcium, characterized by a release profile wherein less than about 90% of the acamprosate calcium is released in 30 minutes pH 6.8.
  • less than about 50% of the acamprosate calcium is released in 120 min at pH 1.2.
  • the formulation is a modified release formulation of acamprosate calcium, characterized by a release profile wherein greater than about 10% of the acamprosate calcium is released in 30 min at pH 1.2.
  • acamprosate calcium is released in 120 min at pH 1.2.
  • the active ingredient comprises about 5 to about 75 w/w% (active ingredient: diluent) of the core.
  • the active ingredient comprises about 5 to about 65 w/w% (active ingredient: diluent) of the core.
  • the active ingredient comprises about 5 to about 60 w/w% (active ingredient: diluent) of the core.
  • the active ingredient comprises about 45 to about 65 w/w% (active ingredient: diluent) of the core.
  • the active ingredient comprises about 50 to about 60 w/w% (active ingredient: diluent) of the core. In a preferred embodiment, the active ingredient comprises about 10 to about 40 w/w% (active ingredient: diluent) of the core.
  • the active ingredient comprises about 20 to about 35 w/w% (active ingredient: diluent) of the core.
  • the active ingredient comprises about 50 w/w% (active ingredient: diluent) of the core.
  • the active ingredient comprises about 60 w/w% (active ingredient: diluent) of the core.
  • the plurality of pellets is contained in a capsule.
  • the capsule contains a unit dose of the active ingredient.
  • the plurality of pellets is contained in a sachet.
  • the sachet contains a unit dose of the active ingredient.
  • the active ingredient is acamprosate calcium and the unit dose is about 100 mg to about 2500 mg.
  • the unit dose is about 333 mg.
  • the sustained release coating is about 1 to about 30 w/w% (sustained release coating: core).
  • the sustained release coating is about 1 to about 10 w/w% (sustained release coating: core).
  • the sustained release coating is about 3 to about 7 w/w%
  • the sustained release coating is about 5 w/w% (sustained release coating: core).
  • the enteric coating is about 25 to about 60 w/w% (enteric coating: core).
  • the enteric coating is about 35 to about 55 w/w% (enteric coating: core).
  • the enteric coating is about 40 to about 50 w/w% (enteric coating: core).
  • the enteric coating is about 50 w/w% (enteric coating: core).
  • the enteric coating is about 40 w/w% (enteric coating: core). In a 17 1 embodiment, the enteric coating is about 25 to about 60 w/w% (enteric coating: core + sustained release coating).
  • the enteric coating is about 35 to about 55 w/w% (enteric coaling: core + sustained release coating).
  • the enteric coating is about 40 to about 50 w/w% (enteric coating: core + sustained release coating).
  • the enteric coating is about 50 w/w% (enteric coating: core + sustained release coating).
  • the enteric coaling is about 40 w/w% (enteric coating: core + sustained release coating).
  • the pharmaceutical formulation further comprises one or more additives selected from the group consisting of: a lubricant, colorant, flow agent, glidant, filler, perfume, flavor, flavor enhancer, or effervescent agent.
  • the pharmaceutical formulation further comprises a mGluR antagonist.
  • the pharmaceutical formulation further comprises an antipsychotic agent.
  • the pharmaceutical formulation further comprises includes a antipsychotic (neuroleptic) medication, a selective serotonin reuptake inhibitor (S SRI), a serotonin-norepinephrine reuptake inhibitor (SNRI), an antidepressant, or an anti-anxiety agent.
  • a antipsychotic (neuroleptic) medication e.g., a selective serotonin reuptake inhibitor (S SRI), a serotonin-norepinephrine reuptake inhibitor (SNRI), an antidepressant, or an anti-anxiety agent.
  • the pharmaceutical formulation further comprises the GABAA agonist bamaluzole, D-Cycloserine, GABA, gabamide, GABOB, gaboxadol, ibotenic acid, isoinpecotic acid, muscimol, phenibut, picamilon, progabide, SL75102, Thiomuscimol, or a positive allosteric modulator that increases the activity of GABA A , such as alcohols, alprazolam, barbiturates, benzodiazepines and nonbenzodiazepines.
  • GABAA agonist bamaluzole D-Cycloserine
  • GABA gabamide
  • GABOB gaboxadol
  • ibotenic acid isoinpecotic acid
  • muscimol phenibut
  • picamilon progabide
  • SL75102 Thiomuscimol
  • a positive allosteric modulator that increases the activity of GABA A , such as alcohols, alprazolam, barbiturates
  • the pharmaceutical formulation further comprises a GABAB receptor agonist, a muscarinic receptor antagonist, a stimulant, a nicotinic receptor agonist, an endocannabinoid receptor antagonist, an AMP A agonist, an antidepressant, an a2-adrenergic agonist, or an anticonvulsant.
  • the present invention provides a method of treating a medical condition in a patient comprising: administering to a patient in need thereof an effective amount of an orally-administrable, pharmaceutical formulation of the present invention.
  • the medical condition is age-related cognitive impairment, Mild Cognitive Impairment (MCI), dementia, Alzheimer's Disease (AD), prodromal AD, posttraumatic stress disorder (PTSD), schizophrenia, bipolar disorder, amyotrophic lateral sclerosis (ALS), cancer-therapy-related cognitive impairment, drug induced or toxin induced cognitive impairment, mental retardation, Parkinson's disease (PD), autism, compulsive behavior, or substance addiction.
  • the medical condition is Alcohol dependence, tinnitus, sleep apnea, Parkinson's disease, levodopa-induced dyskinesias in Parkinson's disease,
  • Alzheimer's disease Huntington's disease, Amyotrophic lateral sclerosis, Cortical spreading depression, migraine, schizophrenia, Anxiety, tardive dyskinesia, spasticity, multiple sclerosis, various types of pain, or binge eating.
  • the medical condition is an Autism Spectrum Disorder, Pervasive Development Disorder-Not Otherwise Specified, Idiopathic Autism, Fragile X Syndrome, Asperger's Syndrome, Rhetl's Syndrome, or Childhood disintegrative disorder as further referenced in Diagnostic and Statistical Manual of Mental Disorders IV.
  • the medical condition is Fragile X Syndrome.
  • the medical condition is a neurotransmission or cognitive disorder that is characterized as a glulamale-GABA imbalance; a disorder characterized with disrupted or dysregulated ERK signaling pathway; or a rasopathy resulting in an abnormality in brain development, learning, memory or cognition.
  • the administering step comprises: sprinkling the pharmaceutical formulation onto food having about pH ⁇ 5.5 prior to ingestion.
  • the food is applesauce or yogurt.
  • a single unit does of the active ingredient is administered 1-4 times per day.
  • the single unit dose is administered 2-3 times per day.
  • Bioavailability refers to the rate and amount of a drug that reaches the systemic circulation of a subject following administration of the drug to the subject and can be determined by evaluating, for example, the plasma or blood concentration-versus-time profile for a drug.
  • Enteric coating refers to a polymer barrier applied (o or on an oral medication in order to protect the drug from the lower pH of the stomach and avoiding dissolution and irritation of the stomach.
  • “Pharmaceutically acceptable” refers to approved or approvable by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopoeia or other generally recognized pharmacopoeia for use in animals, and more particularly in humans.
  • “Pharmaceutically acceptable salt” refers to a salt of a compound, which possesses the desired pharmacological activity of the parent compound.
  • the active ingredient is acamprosate
  • the preferred salt is the calcium salt.
  • “Pharmaceutical composition” refers to at least one active ingredient and at least one pharmaceutically acceptable vehicle with which at least one active ingredient is administered to a subject.
  • Salt refers to a chemical compound consisting of an assembly of cations and anions. Salts of a compound of the present disclosure include stoichiometric and non -stoichiometric forms of the salt. In certain embodiments, because of its potential use in medicine, salts of an active ingredient are pharmaceutically acceptable salts.
  • Spray formulation refers to enteric-coated beads or pellets which can be spherical in shape and is currently defined by the FDA to be .82 mm to 3.04 mm (+ or - 10% variation) in size and can be administered orally with food with or without chewing.
  • Sprinkles can be manufactured in several shapes such as cylindrical, cylindrical with round ends, dumb-bell, ellipsoid or spherical in shape. See “Guidance for Industry Size of Beads in Drug Products Labeled for Sprinkle, " U.S. Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER) May 2012 CMC Rev. 1.
  • “Sachet” is a small flexible package made by bonding, e.g., two layers together on all four sides.
  • the term often refers to single-use, sealed, flexible aluminum pouches which contains a dose of the formulation of which could be presented as a liquid, powder, cream, paste or granule.
  • Subject refers to a mammal, for example, a human.
  • sustained release refers to release of a compound from a dosage form of a pharmaceutical composition at a rate effective to achieve a therapeutic or prophylactic concentration of the compound, or active metabolite thereof, in the systemic circulation of a subject over a prolonged period of time relative to that achieved by administration of an immediate release formulation of the same compound by the same route of administration, hi some embodiments, release of a compound occurs over a time period of at least about 4 hours, at least about 8 hours, at least about 12 hours, at least about 16 hours, at least about 20 hours, and in some embodiments, at least about 24 hours.
  • Treating” or “treatment” of any disease refers to arresting or ameliorating a disease or at least one of the clinical symptoms of a disease or disorder, reducing the risk of acquiring a disease or at least one of the clinical symptoms of a disease, reducing the development of a disease or at least one of the clinical symptoms of the disease or reducing the risk of developing a disease or at least one of the clinical symptoms of a disease.
  • Treating also refers to inhibiting the disease, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both, and to inhibiting at least one physical parameter that may or may not be discernible to the subject.
  • treating refers to delaying the onset of the disease or at least one or more symptoms thereof in a subject which may be exposed to or predisposed to a disease or disorder even though that subject does not yet experience or display symptoms of the disease.
  • “Therapeutically effective amount” refers to the amount of a compound that, when administered to a subject for treating a disease, or at least one of the clinical symptoms of a disease, is sufficient to affect such treatment of the disease or symptom thereof.
  • therapeutically effective amount may vary depending, for example, on the compound, the disease and/or symptoms of the disease, severity of the disease and/or symptoms of the disease or disorder, the age, weight, and/or health of the subject to be treated, and the judgment of the prescribing physician. An appropriate amount in any given instance may be ascertained by those skilled in the art or capable of determination by routine experimentation.
  • “Therapeutically effective dose” refers to a dose that provides effective treatment of a disease or disorder in a subject.
  • a therapeutically effective dose may vary from compound to compound, and from subject to subject, and may depend upon factors such as the condition of the subject and the route of delivery.
  • a therapeutically effective dose may be determined in accordance with routine pharmacological procedures known to those skilled in the art.
  • the pellet cores of the invention comprise one of more active ingredients.
  • the product may include, for example, a single dosage form unit that includes, or consists essentially of, acamprosate and at least one second medication.
  • the active ingredient includes at least one homotaurine analog selected from the group consisting of: an acetylaminopropane sulfonate, an acetyl am inopropane sulfonate salt, taltrimide, and tauromustine.
  • a preferred active ingredient is acamprosate.
  • Acamprosate and its derivatives can be used as single agents or in combination therapy with mGluR antagonists to treat Autism Spectrum Disorders, Fragile X syndrome, Down's syndrome and other forms of mental retardation.
  • Suitable mGluR antagonists are Group I mGluR antagonists including, for example, 2-methyl-6-(phenylethynyl)-pyridine (MPEP), (E)-6-methyl-2-styrylpyridine (SIB 1893), 6-methyl-2-(phenylazo )-3-pyridinol and a-methyl-4-carboxyphenylglycine (MCPG).
  • mGluR antagonists that can be used as single agents or in combination in the pellet core are described in U.S. Patent Nos. 6,890,931 and 6,916,821.
  • suitable mGluR antagonists are mGluR5 antagonists described in WO 01 1661 13, WO 01 132632, WO 01/14390, WO 01 108705, WO 01/05963, WO 01/02367, WO 01/02342, WO 01 102340, WO 00/20001, WO 5 00173283, WO 00/69816, WO 00/63166, WO 00/26199, WO 00/26198, EP-A-0807621 , WO 99/54280, WO 99/44639, WO 99/26927, WO 99108678, WO 99102497, WO 98/45270, WO 98/34907, WO 97/48399, WO 97/48400, WO 97/48409, WO
  • Antipsychotic agents to treat Fragile X syndrome, Down's syndrome and other forms of mental retardation, or autism.
  • Antipsychotic agents including atypical antipsychotic compounds for use in combination treatment can include, for example, abaperidone.acetophenazine maleate, alentemol hydrobromide, alpertine, amisulpride, aripiprazole 5 azaperone, batelapine maleate, benperidol, benzindopyrine hydrochloride, brofoxine.bromperidol, butaclamol hydrochloride, butaperazine, carphenazine maleate, carvolToline hydrochloride, chlorpromazine, chlorprothixene, cinperene, cintriamide, clomacran phosphate, clopenthixol, clopimozide, clopipazan mesylate, cloroperone
  • Other compounds can be used in combination therapy with acamprosate or in singly in the pellet core that include a antipsychotic (neuroleptic) medication, a selective serotonin reuptake inhibitor (SSRI), a serotonin-norepinephrine reuptake inhibitor (SNRI), an antidepressant, an anti-anxiety medication, or the like.
  • a antipsychotic (neuroleptic) medication a selective serotonin reuptake inhibitor (SSRI), a serotonin-norepinephrine reuptake inhibitor (SNRI), an antidepressant, an anti-anxiety medication, or the like.
  • the antipsychotic medication can be, for example, a first or a second generation antipsychotic.
  • the first or a second generation antipsychotic can be for example, one or more of thioridazine, chlorprornazine, thiothixene, trifluoperazine, fluphenazine, haloperidol, perphenazine, loxapine, molindone,
  • the SSRI or SNRI can be, for example, one or more of citalopram, desvenlafaxine, doxepin, duloxetine, escitalopram, fluoxetine, fluvoxamine, milnacipran, paroxetine, sertraline, venlafaxine, and the like.
  • compounds of acamprosate calcium can be used singly or in combination therapy with at least one compound selected from the group consisting of the GABA A agonist bamaluzole, D-Cycloserine, GABA, gabamide, GABOB, gaboxadol, ibotenic acid, isoinpecotic acid, muscimol, phenibut, picamilon, progabide, SL75102,
  • Thiornuscimol, and positive allosteric modulators thai increase (he activity of GABA A such as alcohols, alprazolam, barbiturates, benzodiazepines and nonbenzodiazepines.
  • compounds of acamprosate calcium can be used singly or in combination therapy with at least one compound selected from the group consisting of a GABAe receptor agonist, a muscarinic receptor antagonist, a stimulant, a nicotinic receptor agonist, an endocannabinoid receptor antagonist, an AMP A agonist, an antidepressant, an a2-adrenergic agonist, or an anticonvulsant to treat Fragile X syndrome, Down's syndrome and other forms of mental retardation, or autism.
  • the GABAB receptor agonist is baclofen, R-baclofen, or a prodrug thereof, for example as disclosed in US Patent Nos.
  • the muscarinic receptor antagonist is atropine, benztropine, biperiden, dicyclomine, ipratroprium, procyclidine, scopolamine, tiotropium, telenzepine or trihexyphenidyl.
  • the stimulant is amantadine, bupropion, atomoxetine, dextroamphetamine, modafinil, caffeine,
  • methylphenidate nicotine, pseudoephedrine, and amphetamine, as well as metabolites, isomers or prodrugs thereof.
  • the active compound is preferably homogeneously dispersed in the core and is released with a delay after the surrounding enteric coating is dissolved.
  • the pellet cores of the invention may comprise any acceptable diluent, such as Acacia, Gelatin, Hydroxypropyl cellulose, Hydroxypropylmethyl cellulose, Mel hylcellu lose, Polyetylene glycol (PEG), Povidone, Sucrose, Starch, or a mixture of any of these. Methylcellulose is a preferred diluent.
  • the pellets of the invention include a coating comprising a release-modifying agent, such as ethyl cellulose, caniauba wax, shellac, or a mixture of any of these.
  • a release-modifying agent such as ethyl cellulose, caniauba wax, shellac, or a mixture of any of these.
  • Ethyl cellulose is preferred.
  • enteric coating of the pellets of the invention should only dissolve after the formulation has left the stomach.
  • Such coatings are disclosed in the prior art (e.g., EP 0 453 001 Al).
  • Preferred enteric coatings according to the invention comprise a methacrylic acid copolymer or raethylhydroxypropylcellulose phthalate.
  • Poly(inethacrylic acid, methyl methacrylates), which are obtainable under the tradenames Eudragit® L or S and have free carboxyl groups as functional groups, are preferred. These polymers are insoluble in the gastric juice, but dissolve in digestive juices above pH 5.5-7 depending on the number of functional carboxyl groups.
  • Poly (methacrylic acid, methyl methacrylate) 1 : 1 (Eudragit® L 100; methacrylic acid copolymer, USP/NF type A) and poly(me(hacrylic acid, methyl methacrylate) 1 :2 (Eudragit® S; methacrylic acid copolymer, USP/NF type B) are particularly preferred.
  • Eudragit® L 100 is the most preferred. Mixtures of the coating materials mentioned, in particular of Eudragit® L and Eudragit® S, can also be used.
  • the pellet formulations of the invention can optionally contain one or more conventional additives such as lubricants, colorants, flow agents, glidants, fillers, perfumes, flavors, flavor enhancers, such as sweeteners (both artificial and natural), and other conventional pharmaceutical additives, e.g., effervescent agents.
  • conventional additives such as lubricants, colorants, flow agents, glidants, fillers, perfumes, flavors, flavor enhancers, such as sweeteners (both artificial and natural), and other conventional pharmaceutical additives, e.g., effervescent agents.
  • lubricants such as adipic acid, magnesium stearate, calcium stearate, zinc stearate, hydrogenated vegetable oils, sodium chloride, sterotex, polyoxyethylene, glyceryl monostearate, talc, polyethylene glycol, sodium benzoate, sodium lauryl sulfate, magnesium lauryl sulfate, sodium stearyl fumarate, light mineral oil and the like may be employed, with sodium stearyl fumarate preferred.
  • Waxy fatty acid esters such as glyceryl behenate, sold as "Compritol” products, can be used.
  • Other useful commercial lubricants include "Stear-O-Wet" and "Myvatex TL". Mixtures are operable.
  • Glidants such as starch, talc, lactose, stearales, dibasic calcium phosphate, magnesium carbonate, magnesium oxide, calcium silicate, Cabosil, Syloid, and silicon dioxide aerogels may be employed.
  • Fillers may be used, for example, to increase the bulk of the sachet.
  • Some of the commonly used fillers are calcium sulfate, both di- and rri-basic; starch; calcium carbonate; microcrystalline cellulose; modified starches, lactose, sucrose; mannitol; and sorbitol.
  • Flavors may be chosen from natural and synthetic flavoring liquids.
  • An illustrative list of such agents includes volatile oils, synthetic flavor oils, flavoring aromatics, oils, liquids, oleoresins and extracts derived from plants, leaves, flowers, fruits, stems and combinations thereof.
  • a non-limiting representative list of these includes citric oils, such a lemon, orange, grape, lime and grapefruit an fruit essences, including apple, pear, peach, grape, strawberry, raspberry, cherry, plum, pineapple, apricot, or other fruit flavors.
  • aldehydes and esters such as benzaldehyde (cherry, almond); citral, i.e., alpha-citral (lemon, lime); neral, i.e., beta-citral (lemon, lime); decanal (orange, lemon); aldehyde C-8 (cirrus fruits); aldehyde C-9 (citrus fruits); aldehyde C-12 (citrus fruits); tolyl aldehyde (cherry, almond); 2,6-dimethyloctanal (green fruit); 2-dodedenal (citrus, mandarin); mixtures thereof and the like.
  • aldehydes and esters such as benzaldehyde (cherry, almond); citral, i.e., alpha-citral (lemon, lime); neral, i.e., beta-citral (lemon, lime); decanal (orange, lemon); aldehyde C-8 (cirrus fruits); aldeh
  • Sweeteners may be chosen from the following non-limiting list: glucose (corn syrup), dextrose, invert sugar, fructose, and mixtures thereof (when not used as a carrier); saccharin and its various salts, such as the sodium salt; dipeptide sweeteners such as aspartame;
  • dihydro-chalcone compounds glycyrrhizin; Sfevia Rebaudiana (Stevioside); chloro derivatives or sucrose such as sucralose; and sugar alcohols such as sorbitol, mannitol, xylilol, and the like.
  • hydrogenated starch hydrolysates and the synthetic sweeteners such as 3,6-dihydro-6-methyl- l -l - l ,2,3-oxathiai!;in-4-one-2,2-dioxide, particularly the potassium salt (acesulfame-K), and sodium and calcium salts thereof.
  • Some embodiments include effervescent agents to aid in masking the objectionable taste of active ingredients, such as vitamins, medicines and/or minerals, etc.
  • active ingredients such as vitamins, medicines and/or minerals, etc.
  • Color additives can be used. Such color additives include food, drug and cosmetic colors (FD&C), drug and cosmetic colors (D&C) or external drug and cosmetic colors (Ext. D&C). These colors are dyes, lakes, and certain natural and derived colorants. Useful lakes include dyes absorbed on aluminum hydroxide or other suitable carriers.
  • compositions provided by the present disclosure may be practiced with dosage forms adapted to provide sustained release of an active ingredient upon oral administration.
  • Sustained release oral dosage forms may be used to release drugs over a prolonged time period and are useful when it is desired that a drug or drug form be delivered to the lower gastrointestinal tract.
  • Sustained release oral dosage forms include any oral dosage form that maintains therapeutic concentrations of a drug in a biological fluid such as the plasma, blood, cerebrospinal fluid, or in a tissue or organ for a prolonged time period.
  • Sustained release oral dosage forms include diffusion-controlled systems such as reservoir devices and matrix devices, dissolution-controlled systems, osmotic systems, and erosion- controlled systems. Sustained release oral dosage forms and methods of preparing the same are well known in the art.
  • an active ingredient may be released from an orally administered dosage form over a sufficient period of time to provide prolonged therapeutic concentrations of an active ingredient in the plasma and/or blood of a sub ject.
  • a dosage form comprising an active ingredient may provide a therapeutically effective concentration of the corresponding drug in the plasma and/or blood of a subject for a continuous time period, e.g., for at least about 4 hours, for at least about 8 hours, for at least about 12 hours, for at least about 16 hours, and in certain embodiments, for at least about 20 hours following oral administration of the dosage form to the subject.
  • the continuous time periods during which a therapeutically effective concentration of the drug is maintained may be the same or different.
  • the continuous period of time during which a therapeutically effective plasma concentration of the drug is maintained may begin shortly after oral administration or Following a time interval.
  • An appropriate dosage of an active ingredient or pharmaceutical composition comprising an active ingredient may be determined according to any one of several well-established protocols. For example, animal studies such as studies using mice, rats, dogs, and/or monkeys may be used to determine an appropriate dose of a pharmaceutical compound. Results from animal studies may be extrapolated to determine doses for use in other species, such as for example, humans. USES OF FORMULATIONS
  • this disclosure is directed to acamprosate oral pellet formulations in the manufacture of a medicament for use in methods of treatment of any neurotransmission or cognitive disorder that is characterized as a glutamate-GABA imbalance, any disorder characterized with disrupted or dysregulated ERK signaling pathway or rasopathies resulting in abnormalities in brain development, learning, memory and cognition.
  • Alzheimer's Disease Alzheimer's Disease, Huntington's Disease, Amyotrophic Lateral Sclerosis, Cortical spreading depression, migraine, schizophrenia, anxiety, tardive dyskinesia, spasticity, multiple sclerosis, various types of pain, or binge eating, subjects having or at risk for age- related cognitive impairment, Mild Cognitive Impairment (MCI), dementia, Alzheimer's Disease(AD), prodromal AD, post-traumatic stress disorder (PTSD), bipolar disorder, amyotrophic lateral sclerosis (ALS), cancer-therapy-related cognitive impairment, drug induced or toxin induced cognitive impairments, compulsive behavior, and substance addiction.
  • MCI Mild Cognitive Impairment
  • AD Alzheimer's Disease
  • prodromal AD post-traumatic stress disorder
  • ALS amyotrophic lateral sclerosis
  • cancer-therapy-related cognitive impairment drug induced or toxin induced cognitive impairments, compulsive behavior, and substance addiction.
  • children with mental retardation, Autism Spectrum Disorders, Down's Syndrome and Fragile X Syndrome can be treated with a formulation of the invention.
  • the children can be treated during infancy (between about 0 to about 1 year of life), childhood (the period of life between infancy and puberty) and during puberty (between about 8 years of life to about 18 years of life).
  • the methods disclosed herein can be used to treat adults (greater than about 18 years of life) having mental retardation, Fragile X Syndrome, Autism Spectrum Disorders and/or Down's Syndrome.
  • Disorders, mental retardation and/or Down's syndrome can be treated by administering to the subjects a formulation of the invention.
  • an active ingredient that will be effective in the treatment of a disease in a subject will depend, in part, on the nature of the condition and can be determined by standard clinical techniques known in the art. In addition, in vitro or in vivo assays may be employed to help identify optimal dosage ranges. A therapeutically effective amount of an active ingredient to be administered may also depend on, among other factors, the subject being treated, the weight of the subject, the severity of the disease, the manner of
  • the unit dose of the drug is generally about 100 mg to about 2500 mg. Preferably, the unit dose is about 200 to about 500 mg (e.g, about 333 mg).
  • the unit dose form is typically administered 1 -4 times per day, preferably 2-3 times per day.
  • the pellet formulations of the invention are typically intended for oral administration.
  • the pellets can be packaged in sachets, which are opened at the time of use, and the drug product sprinkled onto food for ingestion.
  • Preferred foods have a pH of less than about 5.5, such as applesauce and yogurt.
  • the pellets can also be encased in a capsule, which can either be taken as such, or the capsule can be opened and the contents sprinkled onto food for ingestion.
  • the target product attributes of the Acamprosate Sprinkles dosage form are summarized in Table 1.
  • the API is known to cause GI irritation and the two current marketed formulations are an enteric-coated tablet, Campral (Campra) is manufactured and marketed in the United States by Forest Laboratories, while Merck KGaA markets it outside the US) and Acampral (Sun Pharmaceuticals).
  • Campral Campra
  • the formulation development approach was to manufacture the pellets sprinkles and coaling with an enteric polymer to avoid or minimize the release of API into the stomach when administered orally.
  • the objectives involved manufacture of pellets of Acamprosate Calcium, with suitable excipient and coating of pellets, to optimize drug release along with shelf stability evaluation of two different sizes/diameters. Sprinkles with two different sizes/diameters were designed with further down-selection to a preferred size/diameter based the drug release profile.
  • Table 3 List of excipients used for Acamprosate calcium sprinkles
  • Acamprosate Calcium sprinkles is shown in Table 4.
  • the formula composition for the manufacture of sprinkles is shown in Table 10.
  • the sprinkles were manufactured by extrusion of wet mass of API+Avicel PH 101 through a mesh size of 1.2 mm.
  • the extruded material was charged into a spheronizer at plate RPM of 2.1 for 10 min to obtain spherical shaped sprinkles.
  • Table 10 Composition of Acamprosate calcium sprinkles (B. No. SF14000746)
  • Table 12 Formula composition for Ethyl cellulose coating (B.No. SFl 4000938)
  • Ethyl cellulose coated sprinkles are further coated with Eudragit L 100 55 polymer for enteric protection.
  • the formula composition for the enteric coating is shown in Table 14, and the manufacturing procedure for the enteric coating is shown in Table 15.
  • Table 14 Formula composition for enteric coating (B.No. SF14000938)
  • the smaller size sprinkles were also manufactured using API and Avice] PH 101 similar to bigger size sprinkles at 1 : 1 mixture.
  • the mesh size for smaller sprinkles was 0.8 mm as compared to 1.2 mm for the larger sprinkles.
  • the Formula composition for the 20% ethyl cellulose sustained release coating is shown in Table 18, and the manufacturing procedure in Table 19.
  • Table 18 Formula composition for Ethyl cellulose coating (B.No. SF 14000999)
  • the drug release was found to be 90% after 5 hours and complete release after the end of 24 hours indicting that 100% drug is present in sprinkles.
  • the sprinkles were then used for enteric coating.
  • the formula composition of the enteric coating is shown in Table 21 , and the manufacturing procedure in Table 22.
  • Table 21 Formula composition for enteric-coating (B.No. SFl 4000999)
  • the sprinkles are intended to be administered in an edible matrix, such as applesauce or yogurt. Most brands of applesauce have a pH in the range of 3-3.6. To determine the drug release in pH 3.0 and 4.5, a dissolution study was conducted on the sprinkles, and the results are shown in Tables 24 and 25.
  • Acamprosate calcium enteric-coated sprinkles were manufactured using #25/30 ASTM sprinkles. Coating was performed with 20% ethyl cellulose (sustained release) followed with 50% enteric coating (Eudragit L 100 55) to achieve the targeted dissolution profile. The same coating procedure was followed as shown in previous section for ethyl cellulose and enteric coating including the formula composition.
  • the batch number for the PK batch is SF 14001404.
  • Acamprosate calcium sprinkles manufactured at two different sizes were found to meet the required target product attributes when compared to marketed products.
  • Ethyl cellulose coating at 20% is required to sustain the release over a period of hours and enteric coating to avoid the release in gastric fluid upon oral administration of sprinkles.
  • Table 280 List of excipients used for Acamprosate calcium sprinkles
  • the sprinkle dosage form of the first generation studies was evaluated in dog PK study wilh formulation composition with 50% drug load acamprosate calcium pellets, with 20% ethyl cellulose coating for sustained release and 50% eudragit coating for enteric protection.
  • the pellets were observed in feces of dog, AUC was lower compared to Campral tablets and Tmax was 12- 16 h in the dog PK study.
  • the further development and optimization of sprinkles was focused on increasing the drug load in pellets, lower the ethyl cellulose percentage to expedite the drug release and optimize/lower the enteric coating.
  • the formula composition for the manufacture of sprinkles is shown in Table 35.
  • the manufacturing procedure for the sprinkles includes an extrusion phase followed spheronization phase, outlined as follows:
  • Step 1 Acamprosate and Avicel PH 101 were mixed in a bowl in 60:40 ratio.
  • Step 2 water was added gradually to prepare a damp mass
  • Step 3 Damp mass was passed through extruder at 100 rpm extruder speed with 0.8 mm single cone screen
  • Step 4 extrudes were further taken for spheronization Spheronization
  • Step 1 2.1 1 mm check plate was used in the spheronization.
  • Step 2 Collected extrudes were added into spheronizer bowl
  • Step 3 Spheronizer was run at 500 rpm (o 800 rpni speed
  • Step 4 5 to 10 mL of water was added drop wise during the spheronization.
  • Step 5 Pellets were further dried in hot air oven.
  • Table 36 Formula composition for Ethyl cellulose coating
  • the ethyl cellulose coated sprinkles are further coated with Eudragit L 100 55 polymer for enteric protection.
  • the formula composition for enteric coaling is shown in Table 38 and the manufacturing procedure in Table 39.
  • the drug release of 5% ethyl cellulose followed with 40% enteric coating sprinkles was found to be 8% in pH 1.2 and the drug release of 5% ethyl cellulose followed with 50% enteric coating was found to be 4% in pH 1.2. However, the drug release in buffer stage was found to be rapid.
  • the pellets were directly added into a petri plate and covered with aluminum foil and pin holes to create open condition and stored at 40 °C/75% RH.
  • the dissolution results of initial and open petri plate study with different ethyl cellulose and Eudragit coating percentages is compiled in Table 41 and the profile is shown in Figure 2-4.
  • Acamprosate calcium pellets in acid stage found to be similar as compared to initial dissolution, which indicates that Eudragit coating is intact and has no influence on faster release.
  • the release rate in buffer stage was similar both at initial and 2 week time point during open exposure.
  • the drug release in buffer stage was found to be faster for 10% ethyl cellulose coaling, indicating (hat higher ethyl cellulose percentage shows faster drug release at higher temperature.
  • Acamprosate calcium sprinkles manufactured with 5% ethyl cellulose and 40% to 50% eudragit coating were found to meet the target product attributes and also were found to be stable for a period of 6 months.
  • Pharmacokinetic study '537 was conducted in beagle dogs at Covance's Madison, Wisconsin facility. This accepted animal model is used to understand how our drug's active ingredient would be absorbed, distributed and eliminated. It should be noted that beagle dogs were used as the animal model for Campral and published research is available for comparison (Kathleen Habemy-FDA Review, Adam Wasserrnan-FDA review and Rliee et al., 2008 a, b). Study '537 allowed us to demonstrate the baseline profile for the existing 333mg enteric-coated tablet across several dose ranges, and at the same time compare a simple "naked" formulation with no excipients where the active ingredient was dissolved in an aqueous solution.
  • the following target product profile for two pedtatric-friendly formulation technologies was developed which could meet the special needs of the Fragile X Syndrome patient population which experiences difficulties taking pills due to oral fixation issues (chewing, gagging and inability to swallow) as well as sensitivities to color, texture and taste while accommodating the challenges of this BCS III compound.
  • Two dosage forms were contemplated in the target product profile for acamprosate calcium; a micro-sized particle and small -particle Sprinkle form, as described in Table 44.
  • Formulation development activities included preformulation profiling, drug excipient compatibility studies, drug product methods development, formulation development and informal stability of developed formulations.
  • a number of micro-size particle prototypes were rendered using by several different methodologies with no prototype meeting the target product profile. The development team was unable to consistently coat the API
  • microparlicles to delay dissolution.
  • the development team considered sprinkles/pellets uniformly coated with sustained release polymers and enteric coated polymers.
  • SF 14000999 was advanced to dog study '752 where the team analyzed the PK. and Tolerabilily profile of SF14000999 in comparison with Campral tablet.
  • This 7 day study dosed the dogs daily at 1,332 mg BID for an aggregate exposure of 2,664 mg per day. All dogs were male non-naive beagle dogs from the Covance stock colony.
  • the animals weighed 9.4 to 12.0 kg and were young adult/adult. Animals were identified with individually numbered cage cards or an implantable microchip identification device (IMID). Animals were selected for test based on overall health and body weight.
  • the study was conducted under fed conditions and animals were given Certified Canine Diet #5007 and food was provided ad libitum. Water was provided fresh daily ad libitum.
  • a second dog study '326 was conducted to determine if the very high drug exposure for SF 14000999 was the cause of the GI disturbance and further causing insufficient time for the sprinkles to release.
  • Study '326 compared SF 14000999 and Campral dosed at 666 mg BID for an aggregate daily exposure of 1 ,332 mg.
  • the protocol was kept relatively the same with the following exceptions; the study was shortened to 4 days and the dose was lessen to 1 ,332 mg per day.
  • Individual doses were administered as a fixed dose of 2 capsules/tablets per animal (666 mg per animal) twice daily, approximately 12 hours apart, for a total dose of 4 capsules/tablets per day ( 1332 mg per animal). The capsule/tablet doses were administered orally, followed by approximately 10 mL of water.
  • Figure 8 shows the Day 1 PK for the 666 mg BID study. The data demonstrated that the exposures (Cmax/AUC) were lower for sprinkle prototype SF 14000999 than Campral.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Zoology (AREA)
  • Physiology (AREA)
  • Nutrition Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Coloring Foods And Improving Nutritive Qualities (AREA)
PCT/US2016/030725 2015-05-04 2016-05-04 Sprinkle formulations of acamprosate WO2016179252A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
US15/569,937 US10709668B2 (en) 2015-05-04 2016-05-04 Sprinkle formulations of acamprosate
SG11201708393UA SG11201708393UA (en) 2015-05-04 2016-05-04 Sprinkle formulations of acamprosate
KR1020177033137A KR20170142180A (ko) 2015-05-04 2016-05-04 아캄프로세이트의 스프링클 제형
JP2017558372A JP7026347B2 (ja) 2015-05-04 2016-05-04 アカンプロサートのスプリンクル製剤
EP16789996.2A EP3291800A4 (en) 2015-05-04 2016-05-04 Sprinkle formulations of acamprosate
AU2016258624A AU2016258624B2 (en) 2015-05-04 2016-05-04 Sprinkle formulations of acamprosate
IL255343A IL255343B1 (en) 2015-05-04 2016-05-04 Essay formulations of acamprosate
ZA2017/08037A ZA201708037B (en) 2015-05-04 2017-11-27 Sprinkle formulations of acamprosate

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201562156842P 2015-05-04 2015-05-04
US62/156,842 2015-05-04
US201562260161P 2015-11-25 2015-11-25
US62/260,161 2015-11-25

Publications (1)

Publication Number Publication Date
WO2016179252A1 true WO2016179252A1 (en) 2016-11-10

Family

ID=57217823

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/030725 WO2016179252A1 (en) 2015-05-04 2016-05-04 Sprinkle formulations of acamprosate

Country Status (10)

Country Link
US (1) US10709668B2 (zh)
EP (1) EP3291800A4 (zh)
JP (2) JP7026347B2 (zh)
KR (1) KR20170142180A (zh)
AU (1) AU2016258624B2 (zh)
HK (1) HK1252003A1 (zh)
IL (1) IL255343B1 (zh)
SG (1) SG11201708393UA (zh)
WO (1) WO2016179252A1 (zh)
ZA (1) ZA201708037B (zh)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018213589A1 (en) * 2017-05-17 2018-11-22 Confluence Pharmaceuticals, Llc Formulations of homotaurines and salts thereof
EP3331518A4 (en) * 2015-08-04 2019-04-03 Confluence Pharmaceuticals, LLC COMBINED THERAPY USING ACAMPROSATE AND D-CYCLOSERIN
US10709668B2 (en) 2015-05-04 2020-07-14 Confluence Pharmaceuticals, Llc Sprinkle formulations of acamprosate
US11065222B2 (en) 2015-02-18 2021-07-20 Aston University Diagnostic assay and treatment for preeclampsia

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022058989A1 (en) * 2020-09-21 2022-03-24 Sun Pharmaceutical Industries Limited Multi-particulate pharmaceutical composition of quetiapine
EP4406479A1 (en) 2021-09-24 2024-07-31 Sumitomo Bakelite Co.Ltd. Brain wave detection electrode, brain wave measurement device, and brain wave measurement method

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110142889A1 (en) * 2009-12-16 2011-06-16 Nod Pharmaceuticals, Inc. Compositions and methods for oral drug delivery
US20120077878A1 (en) * 2010-09-28 2012-03-29 Depomed, Inc. Gastric retentive dosage forms for extended release of acamprosate into the upper gastrointestinal tract
US20130143867A1 (en) * 2011-12-02 2013-06-06 Sychroneuron Inc. Acamprosate formulations, methods of using the same, and combinations comprising the same

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6514530B2 (en) 1997-09-09 2003-02-04 Alza Corporation Dosage form comprising means for changing drug delivery shape
EP1844769A3 (en) 1998-04-14 2010-02-10 The General Hospital Corporation Methods for treating neuropsychiatric disorders
US20050042277A1 (en) * 2003-07-17 2005-02-24 Irukulla Srinivas Pharmaceutical compositions having a swellable coating
DE102007009243A1 (de) 2007-02-22 2008-09-18 Evonik Röhm Gmbh Pellets mit einer Wirkstoff-Matrix und einem Polymerüberzug, sowie ein Verfahren zur Herstellung der Pellets
JP2010532331A (ja) 2007-07-05 2010-10-07 アンセルム(アンスチチュ ナショナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシュ メディカル) 抗痙攣医薬組成物
JP5688035B2 (ja) 2009-02-12 2015-03-25 インディアナ・ユニバーシティ・リサーチ・アンド・テクノロジー・コーポレーション 合併性および特発性自閉症を含む発達障害を治療するための物質および方法
US20100216805A1 (en) 2009-02-25 2010-08-26 Braincells, Inc. Modulation of neurogenesis using d-cycloserine combinations
SG192968A1 (en) 2011-03-01 2013-09-30 Pharnext Baclofen and acamprosate based therapy of neurogical disorders
ES2644942T3 (es) * 2011-03-23 2017-12-01 Ironshore Pharmaceuticals & Development, Inc. Métodos y composiciones para el tratamiento del trastorno por déficit de atención
KR20130002292A (ko) 2011-06-28 2013-01-07 주식회사 비보존 다중 타겟팅의 상승 효과를 유발하는 유효물질의 조합 및 그 용도
WO2014197744A1 (en) 2013-06-05 2014-12-11 Synchroneuron, Inc. Acamprosate formulations, methods of using the same, and combinations comprising the same
WO2015121218A1 (en) 2014-02-11 2015-08-20 Pharnext Combination of baclofen, acamprosate and medium chain triglycerides for the treatment of neurological disorders
IL255343B1 (en) 2015-05-04 2024-06-01 Confluence Pharmaceuticals Llc Essay formulations of acamprosate

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110142889A1 (en) * 2009-12-16 2011-06-16 Nod Pharmaceuticals, Inc. Compositions and methods for oral drug delivery
US20120077878A1 (en) * 2010-09-28 2012-03-29 Depomed, Inc. Gastric retentive dosage forms for extended release of acamprosate into the upper gastrointestinal tract
US20130143867A1 (en) * 2011-12-02 2013-06-06 Sychroneuron Inc. Acamprosate formulations, methods of using the same, and combinations comprising the same

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
"Ethocel Premium Polymers For Pharmaceutical Applications.", DOW, October 1998 (1998-10-01), pages 1 - 12, XP055006766 *
"Matenal Safety Data Sheet Avicel PH Microcrystalline Cellulose.", FMC BIOPOLYMER, January 2008 (2008-01-01), pages 1 - 9, XP055329476 *
See also references of EP3291800A4 *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11065222B2 (en) 2015-02-18 2021-07-20 Aston University Diagnostic assay and treatment for preeclampsia
US11980600B2 (en) 2015-02-18 2024-05-14 Mirzyme Therapeutics Limited Diagnostic assay and treatment for preeclampsia
US10709668B2 (en) 2015-05-04 2020-07-14 Confluence Pharmaceuticals, Llc Sprinkle formulations of acamprosate
EP3331518A4 (en) * 2015-08-04 2019-04-03 Confluence Pharmaceuticals, LLC COMBINED THERAPY USING ACAMPROSATE AND D-CYCLOSERIN
WO2018213589A1 (en) * 2017-05-17 2018-11-22 Confluence Pharmaceuticals, Llc Formulations of homotaurines and salts thereof
JP2020519669A (ja) * 2017-05-17 2020-07-02 コンフルーエンス ファーマシューティカルズ,エルエルシー ホモタウリンおよびその塩の製剤
EP3624788A4 (en) * 2017-05-17 2021-03-03 Confluence Pharmaceuticals, LLC FORMULATIONS OF HOMOTAURINS AND THEIR SALTS
AU2018269557B2 (en) * 2017-05-17 2021-06-03 Confluence Pharmaceuticals, Llc Formulations of homotaurines and salts thereof
AU2021225172B2 (en) * 2017-05-17 2023-04-06 Confluence Pharmaceuticals, Llc Formulations Of Homotaurines And Salts Thereof
JP7499539B2 (ja) 2017-05-17 2024-06-14 コンフルーエンス ファーマシューティカルズ,エルエルシー ホモタウリンおよびその塩の製剤
US12042566B2 (en) 2017-05-17 2024-07-23 Confluence Pharmaceuticals, Llc Formulations of homotaurines and salts thereof

Also Published As

Publication number Publication date
HK1252003A1 (zh) 2019-05-10
KR20170142180A (ko) 2017-12-27
AU2016258624A1 (en) 2017-11-02
IL255343A0 (en) 2017-12-31
EP3291800A1 (en) 2018-03-14
US20190046458A1 (en) 2019-02-14
AU2016258624B2 (en) 2021-07-29
JP2021155434A (ja) 2021-10-07
JP2018515506A (ja) 2018-06-14
EP3291800A4 (en) 2018-09-26
IL255343B1 (en) 2024-06-01
SG11201708393UA (en) 2017-11-29
US10709668B2 (en) 2020-07-14
ZA201708037B (en) 2020-03-25
JP7026347B2 (ja) 2022-02-28

Similar Documents

Publication Publication Date Title
AU2016258624B2 (en) Sprinkle formulations of acamprosate
ES2922387T3 (es) Microparticulas recubiertas de liberacion modificada de al menos un principio activo y forma galenica oral que las comprende
US10512621B2 (en) Methods of treating posttraumatic stress disorder with acamprosate salts
CN102458360A (zh) 含有高剂量和低剂量药物的组合的口腔崩解片组合物
US20150250746A1 (en) Acamprosate formulations, methods of using the same, and combinations comprising the same
TW200427448A (en) Controlled release preparations comprising tramadol and topiramate
WO2008008592A2 (en) Multi-particulate formulation having tramadol in immediate and controlled release form
JP7499539B2 (ja) ホモタウリンおよびその塩の製剤
EP2785337B1 (en) Acamprosate formulations, methods of using the same, and combinations comprising the same
CA1218603A (en) Sustained release method and product
Patel Formulation and evaluation of ranolazine sustained release tablets by hot melt coating technique
WO2023126967A1 (en) Oral films of antipruritic drugs
US20150238615A1 (en) Denaturants for sympathomimetic amines
DIHYDROCHLORIDE Reg. No. 26106506
TR201613529A1 (tr) Li̇pi̇t düşürücü etki̇ gösteren farmasöti̇k terki̇pler
Joshi et al. INTERNATIONAL JOURNAL OF UNIVERSAL PHARMACY AND BIO SCIENCES
Trivedi In situ forming oral controlled release drug delivery formulations (ISFOF)

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16789996

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 11201708393U

Country of ref document: SG

WWE Wipo information: entry into national phase

Ref document number: 255343

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2017558372

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2016258624

Country of ref document: AU

Date of ref document: 20160504

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20177033137

Country of ref document: KR

Kind code of ref document: A